1
|
Moreno-Vargas LM, Prada-Gracia D. Exploring the Chemical Features and Biomedical Relevance of Cell-Penetrating Peptides. Int J Mol Sci 2024; 26:59. [PMID: 39795918 PMCID: PMC11720145 DOI: 10.3390/ijms26010059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 01/13/2025] Open
Abstract
Cell-penetrating peptides (CPPs) are a diverse group of peptides, typically composed of 4 to 40 amino acids, known for their unique ability to transport a wide range of substances-such as small molecules, plasmid DNA, small interfering RNA, proteins, viruses, and nanoparticles-across cellular membranes while preserving the integrity of the cargo. CPPs exhibit passive and non-selective behavior, often requiring functionalization or chemical modification to enhance their specificity and efficacy. The precise mechanisms governing the cellular uptake of CPPs remain ambiguous; however, electrostatic interactions between positively charged amino acids and negatively charged glycosaminoglycans on the membrane, particularly heparan sulfate proteoglycans, are considered the initial crucial step for CPP uptake. Clinical trials have highlighted the potential of CPPs in diagnosing and treating various diseases, including cancer, central nervous system disorders, eye disorders, and diabetes. This review provides a comprehensive overview of CPP classifications, potential applications, transduction mechanisms, and the most relevant algorithms to improve the accuracy and reliability of predictions in CPP development.
Collapse
|
2
|
Xu X, Zhu N, Zheng J, Peng Y, Zeng MS, Deng K, Duan C, Yuan Y. EBV abortive lytic cycle promotes nasopharyngeal carcinoma progression through recruiting monocytes and regulating their directed differentiation. PLoS Pathog 2024; 20:e1011934. [PMID: 38206974 PMCID: PMC10846743 DOI: 10.1371/journal.ppat.1011934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 02/06/2024] [Accepted: 01/01/2024] [Indexed: 01/13/2024] Open
Abstract
Epstein-Barr virus (EBV) is associated with several types of human cancer including nasopharyngeal carcinoma (NPC). The activation of EBV to the lytic cycle has been observed in advanced NPC and is believed to contribute to late-stage NPC development. However, how EBV lytic cycle promotes NPC progression remains elusive. Analysis of clinical NPC samples indicated that EBV reactivation and immunosuppression were found in advanced NPC samples, as well as abnormal angiogenesis and invasiveness. To investigate the role of the EBV lytic cycle in tumor development, we established a system that consists of two NPC cell lines, respectively, in EBV abortive lytic cycle and latency. In a comparative analysis using this system, we found that the NPC cell line in EBV abortive lytic cycle exhibited the superior chemotactic capacity to recruit monocytes and polarized their differentiation toward tumor-associated macrophage (TAM)-like phenotype and away from DCs, compared to EBV-negative or EBV-latency NPC cells. EBV-encoded transcription activator ZTA is responsible for regulating monocyte chemotaxis and TAM phenotype by up-regulating the expression of GM-CSF, IL-8, and GRO-α. As a result, TAM induced by EBV abortive lytic cycle promotes NPC angiogenesis, invasion, and migration. Overall, this study elucidated the role of the EBV lytic life cycle in the late development of NPC and revealed a mechanism underlying the ZTA-mediated establishment of the tumor microenvironment (TME) that promotes NPC late-stage progression.
Collapse
Affiliation(s)
- Xiaoting Xu
- Laboratory of Clinical, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Nannan Zhu
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Junming Zheng
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yingying Peng
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Kai Deng
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Chaohui Duan
- Laboratory of Clinical, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yan Yuan
- Institute for Advanced Medical Research, Shandong University, Jinan, China
| |
Collapse
|
3
|
Pascolutti R, Yeturu L, Philippin G, Costa Borges S, Dejob M, Santiago-Raber ML, Derouazi M. ATP128 Clinical Therapeutic Cancer Vaccine Activates NF-κB and IRF3 Pathways through TLR4 and TLR2 in Human Monocytes and Dendritic Cells. Cancers (Basel) 2022; 14:cancers14205134. [PMID: 36291919 PMCID: PMC9600632 DOI: 10.3390/cancers14205134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022] Open
Abstract
The use of cancer vaccines is a promising therapeutic strategy able to stimulate anti-tumor immunity by inducing both humoral and cellular immunity. In this study, antigen presenting cells play a key role by inducing a strong activation of the T cell-mediated adaptive immune response, essential for the anti-tumor potential of cancer vaccines. The first human candidate vaccine created from the KISIMA platform, ATP128, bears three tumor-associated antigens highly expressed in colorectal cancer tissues. At the N-terminus, the cell-penetrating peptide allows the antigen delivery inside the cell and, together with the TLR agonist-derived peptide at the C-terminus, ensures the activation of the monocyte-derived dendritic cells. Here, we show that ATP128 leads to both NF-κB and IRF3 pathway activation, with subsequent pro-inflammatory cytokines and type I Interferon release, as well as an increase in the expression of costimulatory molecules, alongside an upregulation of MHC class I molecules. This cellular immune response involves TLR2 and TLR4, for both membrane and intracellular signaling. We demonstrated an endocytic component in ATP128’s activity by combining the use of a variant of ATP128 lacking the cell-penetrating peptide with endocytosis inhibitors. Importantly, this internalization step is detemined essential for the activation of the IRF3 pathway. This study validates the design of the self-adjuvanting ATP128 vaccine for cancer immunotherapy.
Collapse
Affiliation(s)
- Roberta Pascolutti
- AMAL Therapeutics, 1205 Geneva, Switzerland
- Boehringer-Ingelheim GmbH, 55216 Ingelheim, Germany
| | - Lakshmi Yeturu
- AMAL Therapeutics, 1205 Geneva, Switzerland
- Boehringer-Ingelheim GmbH, 55216 Ingelheim, Germany
| | - Géraldine Philippin
- AMAL Therapeutics, 1205 Geneva, Switzerland
- Boehringer-Ingelheim GmbH, 55216 Ingelheim, Germany
| | - Stéphane Costa Borges
- AMAL Therapeutics, 1205 Geneva, Switzerland
- Boehringer-Ingelheim GmbH, 55216 Ingelheim, Germany
| | - Magali Dejob
- AMAL Therapeutics, 1205 Geneva, Switzerland
- Boehringer-Ingelheim GmbH, 55216 Ingelheim, Germany
| | - Marie-Laure Santiago-Raber
- AMAL Therapeutics, 1205 Geneva, Switzerland
- Boehringer-Ingelheim GmbH, 55216 Ingelheim, Germany
- Correspondence: (M.-L.S.-R.); (M.D.)
| | - Madiha Derouazi
- AMAL Therapeutics, 1205 Geneva, Switzerland
- Boehringer-Ingelheim GmbH, 55216 Ingelheim, Germany
- Correspondence: (M.-L.S.-R.); (M.D.)
| |
Collapse
|
4
|
Nečasová I, Stojaspal M, Motyčáková E, Brom T, Janovič T, Hofr C. Transcriptional regulators of human oncoviruses: structural and functional implications for anticancer therapy. NAR Cancer 2022; 4:zcac005. [PMID: 35252867 PMCID: PMC8892037 DOI: 10.1093/narcan/zcac005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 02/04/2022] [Accepted: 02/15/2022] [Indexed: 11/26/2022] Open
Abstract
Transcription is often the first biosynthetic event of viral infection. Viruses produce preferentially viral transcriptional regulators (vTRs) essential for expressing viral genes and regulating essential host cell proteins to enable viral genome replication. As vTRs are unique viral proteins that promote the transcription of viral nucleic acid, vTRs interact with host proteins to suppress detection and immune reactions to viral infection. Thus, vTRs are promising therapeutic targets that are sequentially and structurally distinct from host cell proteins. Here, we review vTRs of three human oncoviruses: HBx of hepatitis B virus, HBZ of human T-lymphotropic virus type 1, and Rta of Epstein-Barr virus. We present three cunningly exciting and dangerous transcription strategies that make viral infections so efficient. We use available structural and functional knowledge to critically examine the potential of vTRs as new antiviral-anticancer therapy targets. For each oncovirus, we describe (i) the strategy of viral genome transcription; (ii) vTRs' structure and binding partners essential for transcription regulation; and (iii) advantages and challenges of vTR targeting in antiviral therapies. We discuss the implications of vTR regulation for oncogenesis and perspectives on developing novel antiviral and anticancer strategies.
Collapse
Affiliation(s)
- Ivona Nečasová
- Institute of Biophysics of the Czech Academy of Sciences, Scientific Incubator, Královopolská 135, Brno 612 65, Czech Republic
| | - Martin Stojaspal
- Institute of Biophysics of the Czech Academy of Sciences, Scientific Incubator, Královopolská 135, Brno 612 65, Czech Republic
| | - Edita Motyčáková
- Institute of Biophysics of the Czech Academy of Sciences, Scientific Incubator, Královopolská 135, Brno 612 65, Czech Republic
| | - Tomáš Brom
- LifeB, Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 753/5, Brno 625 00, Czech Republic
| | - Tomáš Janovič
- LifeB, Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 753/5, Brno 625 00, Czech Republic
| | - Ctirad Hofr
- Institute of Biophysics of the Czech Academy of Sciences, Scientific Incubator, Královopolská 135, Brno 612 65, Czech Republic
| |
Collapse
|
5
|
Santisteban-Espejo A, Perez-Requena J, Atienza-Cuevas L, Moran-Sanchez J, Fernandez-Valle MDC, Bernal-Florindo I, Romero-Garcia R, Garcia-Rojo M. Prognostic Role of the Expression of Latent-Membrane Protein 1 of Epstein–Barr Virus in Classical Hodgkin Lymphoma. Viruses 2021; 13:v13122523. [PMID: 34960792 PMCID: PMC8706848 DOI: 10.3390/v13122523] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 11/16/2022] Open
Abstract
The prognostic impact of the presence of Epstein–Barr virus (EBV) in classical Hodgkin lymphoma (cHL) is controversial. Previous studies reported heterogeneous results, rendering difficult the clinical validation of EBV as a prognostic biomarker in this lymphoma. The objective of this study was to evaluate the survival impact of the expression of EBV Latent-Membrane Protein 1 (EBV-LMP1) in tumoral Hodgkin–Reed–Sternberg (HRS) cells of primary diagnostic samples of cHL. Formalin-Fixed Paraffin-Embedded (FFPE) lymph node samples from 88 patients with cHL were analyzed. Patients were treated with the standard first-line chemotherapy (CT) with Adriamycin, Bleomycin, Vinblastine and Dacarbazine (ABVD) followed by radiotherapy. The Kaplan–Meier method and the Cox proportional hazards model were used for carrying out the survival analysis. In order to investigate whether the influence of EBV was age-dependent, analyses were performed both for patients of all ages and for age-stratified subgroups. In bivariate analysis, the expression of EBV was associated with older age (p = 0.011), mixed cellularity subtype cHL (p < 0.001) and high risk International Prognostic Score (IPS) (p = 0.023). Overall survival (OS) and progression-free survival (PFS) were associated with the presence of bulky disease (p = 0.009) and advanced disease at diagnosis (p = 0.016). EBV-positive cases did not present a significantly lower OS and PFS in comparison with EBV-negative cases, for all ages and when stratifying for age. When adjusted for covariates, absence of bulky disease at diagnosis (HR: 0.102, 95% CI: 0.02–0.48, p = 0.004) and limited disease stages (I–II) (HR: 0.074, 95% CI: 0.01–0.47, p = 0.006) were associated with a significant better OS. For PFS, limited-disease stages also retained prognostic impact in the multivariate Cox regression (HR: 0.145, 95% CI: 0.04–0.57, p = 0.006). These results are of importance as the early identification of prognostic biomarkers in cHL is critical for guiding and personalizing therapeutic decisions. The prognostic role of EBV in cHL could be modulated by the type of CT protocol employed and interact with the rest of presenting features.
Collapse
Affiliation(s)
- Antonio Santisteban-Espejo
- Department of Pathology, Puerta del Mar University Hospital, 11009 Cadiz, Spain; (J.P.-R.); (L.A.-C.); (M.G.-R.)
- Institute of Research and Innovation in Biomedical Sciences of the Province of Cadiz (INiBICA), 11009 Cadiz, Spain; (I.B.-F.); (R.R.-G.)
- Department of Medicine, Faculty of Medicine, University of Cadiz, 11003 Cadiz, Spain;
- Correspondence:
| | - Jose Perez-Requena
- Department of Pathology, Puerta del Mar University Hospital, 11009 Cadiz, Spain; (J.P.-R.); (L.A.-C.); (M.G.-R.)
| | - Lidia Atienza-Cuevas
- Department of Pathology, Puerta del Mar University Hospital, 11009 Cadiz, Spain; (J.P.-R.); (L.A.-C.); (M.G.-R.)
| | - Julia Moran-Sanchez
- Department of Medicine, Faculty of Medicine, University of Cadiz, 11003 Cadiz, Spain;
- Department of Hematology and Hemotherapy, Puerta del Mar University Hospital, 11009 Cadiz, Spain;
| | | | - Irene Bernal-Florindo
- Institute of Research and Innovation in Biomedical Sciences of the Province of Cadiz (INiBICA), 11009 Cadiz, Spain; (I.B.-F.); (R.R.-G.)
| | - Raquel Romero-Garcia
- Institute of Research and Innovation in Biomedical Sciences of the Province of Cadiz (INiBICA), 11009 Cadiz, Spain; (I.B.-F.); (R.R.-G.)
| | - Marcial Garcia-Rojo
- Department of Pathology, Puerta del Mar University Hospital, 11009 Cadiz, Spain; (J.P.-R.); (L.A.-C.); (M.G.-R.)
- Institute of Research and Innovation in Biomedical Sciences of the Province of Cadiz (INiBICA), 11009 Cadiz, Spain; (I.B.-F.); (R.R.-G.)
| |
Collapse
|
6
|
Faure C, Djerbi-Bouillié R, Domingot A, Bouzinba-Segard H, Taouji S, Saidi Y, Bernard S, Carallis F, Rothe-Walther R, Lenormand JL, Chevet E, Bourdoulous S. Allosteric Inhibition of HER2 by Moesin-Mimicking Compounds Targets HER2-Positive Cancers and Brain Metastases. Cancer Res 2021; 81:5464-5476. [PMID: 34493594 DOI: 10.1158/0008-5472.can-21-0162] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 07/27/2021] [Accepted: 09/02/2021] [Indexed: 11/16/2022]
Abstract
Therapies targeting the tyrosine kinase receptor HER2 have significantly improved survival of patients with HER2+ cancer. However, both de novo and acquired resistance remain a challenge, particularly in the brain metastatic setting. Here we report that, unlike other HER tyrosine kinase receptors, HER2 possesses a binding motif in its cytosolic juxtamembrane region that allows interaction with members of the Ezrin/Radixin/Moesin (ERM) family. Under physiologic conditions, this interaction controls the localization of HER2 in ERM-enriched domains and stabilizes HER2 in a catalytically repressed state. In HER2+ breast cancers, low expression of Moesin correlated with increased HER2 expression. Restoring expression of ERM proteins in HER2+ breast cancer cells was sufficient to revert HER2 activation and inhibit HER2-dependent proliferation. A high-throughput assay recapitulating the HER2-ERM interaction allowed for screening of about 1,500 approved drugs. From this screen, we found Zuclopenthixol, an antipsychotic drug that behaved as a Moesin-mimicking compound, because it directly binds the juxtamembrane region of HER2 and specifically inhibits HER2 activation in HER2+ cancers, as well as activation of oncogenic mutated and truncated forms of HER2. Zuclopenthixol efficiently inhibited HER2+ breast tumor progression in vitro and in vivo and, more importantly, showed significant activity on HER2+ brain tumor progression. Collectively, these data reveal a novel class of allosteric HER2 inhibitors, increasing the number of approaches to consider for intervention on HER2+ breast cancers and brain metastases. SIGNIFICANCE: This study demonstrates the functional role of Moesin in maintaining HER2 in a catalytically repressed state and provides novel therapeutic approaches targeting HER2+ breast cancers and brain metastasis using Moesin-mimicking compounds.
Collapse
Affiliation(s)
- Camille Faure
- Université de Paris, Institut Cochin, Inserm, CNRS, Paris, France.
| | | | - Anaïs Domingot
- Université de Paris, Institut Cochin, Inserm, CNRS, Paris, France
| | | | - Saïd Taouji
- Inserm, Université de Bordeaux, Institut Bergonié, Bordeaux, France
| | - Yanis Saidi
- Université de Paris, Institut Cochin, Inserm, CNRS, Paris, France
| | - Sandra Bernard
- Université de Paris, Institut Cochin, Inserm, CNRS, Paris, France
| | | | - Romy Rothe-Walther
- TIMC-IMAG Laboratory, CNRS, Université Joseph Fourier, UFR de Médecine, La Tronche, France
| | - Jean-Luc Lenormand
- TIMC-IMAG Laboratory, CNRS, Université Joseph Fourier, UFR de Médecine, La Tronche, France
| | - Eric Chevet
- Inserm, Université de Bordeaux, Institut Bergonié, Bordeaux, France
| | | |
Collapse
|
7
|
Min S, Kim K, Ku S, Park J, Seo J, Roh S. Newly synthesized peptide, Ara‐27, exhibits significant improvement in cell‐penetrating ability compared to conventional peptides. Biotechnol Prog 2020; 36:e3014. [DOI: 10.1002/btpr.3014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 03/17/2020] [Accepted: 04/21/2020] [Indexed: 11/10/2022]
Affiliation(s)
- Sol Min
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, School of Dentistry Seoul National University Seoul Korea
| | - Kichul Kim
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, School of Dentistry Seoul National University Seoul Korea
| | - Seockmo Ku
- Fermentation Science Program, School of Agriculture, College of Basic and Applied Sciences Middle Tennessee State University Murfreesboro Tennessee USA
| | - Jeong‐Yoon Park
- The Spine and Spinal Cord Institute, Department of Neurosurgery, Gangnam Severance Hospital Yonsei University College of Medicine Seoul Republic of Korea
| | - Jeongmin Seo
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, School of Dentistry Seoul National University Seoul Korea
- Biomedical Research Institute NeoRegen Biotech Co., Ltd. Gyeonggi‐do Korea
| | - Sangho Roh
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, School of Dentistry Seoul National University Seoul Korea
| |
Collapse
|
8
|
Germini D, Sall FB, Shmakova A, Wiels J, Dokudovskaya S, Drouet E, Vassetzky Y. Oncogenic Properties of the EBV ZEBRA Protein. Cancers (Basel) 2020; 12:E1479. [PMID: 32517128 PMCID: PMC7352903 DOI: 10.3390/cancers12061479] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 12/14/2022] Open
Abstract
Epstein Barr Virus (EBV) is one of the most common human herpesviruses. After primary infection, it can persist in the host throughout their lifetime in a latent form, from which it can reactivate following specific stimuli. EBV reactivation is triggered by transcriptional transactivator proteins ZEBRA (also known as Z, EB-1, Zta or BZLF1) and RTA (also known as BRLF1). Here we discuss the structural and functional features of ZEBRA, its role in oncogenesis and its possible implication as a prognostic or diagnostic marker. Modulation of host gene expression by ZEBRA can deregulate the immune surveillance, allow the immune escape, and favor tumor progression. It also interacts with host proteins, thereby modifying their functions. ZEBRA is released into the bloodstream by infected cells and can potentially penetrate any cell through its cell-penetrating domain; therefore, it can also change the fate of non-infected cells. The features of ZEBRA described in this review outline its importance in EBV-related malignancies.
Collapse
Affiliation(s)
- Diego Germini
- CNRS UMR9018, Université Paris-Saclay, Institut Gustave Roussy, 94805 Villejuif, France; (D.G.); (F.B.S.); (A.S.); (J.W.); (S.D.)
| | - Fatimata Bintou Sall
- CNRS UMR9018, Université Paris-Saclay, Institut Gustave Roussy, 94805 Villejuif, France; (D.G.); (F.B.S.); (A.S.); (J.W.); (S.D.)
- Laboratory of Hematology, Aristide Le Dantec Hospital, Cheikh Anta Diop University, Dakar 12900, Senegal
| | - Anna Shmakova
- CNRS UMR9018, Université Paris-Saclay, Institut Gustave Roussy, 94805 Villejuif, France; (D.G.); (F.B.S.); (A.S.); (J.W.); (S.D.)
| | - Joëlle Wiels
- CNRS UMR9018, Université Paris-Saclay, Institut Gustave Roussy, 94805 Villejuif, France; (D.G.); (F.B.S.); (A.S.); (J.W.); (S.D.)
| | - Svetlana Dokudovskaya
- CNRS UMR9018, Université Paris-Saclay, Institut Gustave Roussy, 94805 Villejuif, France; (D.G.); (F.B.S.); (A.S.); (J.W.); (S.D.)
| | - Emmanuel Drouet
- CIBB-IBS UMR 5075 Université Grenoble Alpes, 38044 Grenoble, France;
| | - Yegor Vassetzky
- CNRS UMR9018, Université Paris-Saclay, Institut Gustave Roussy, 94805 Villejuif, France; (D.G.); (F.B.S.); (A.S.); (J.W.); (S.D.)
- Koltzov Institute of Developmental Biology, 117334 Moscow, Russia
| |
Collapse
|
9
|
Abstract
The integration of drugs into nanocarriers favorably altered their pharmacodynamics and pharmacokinetics compared to free drugs, and increased their therapeutic index. However, selective cellular internalization in diseased tissues rather than normal tissues still presents a formidable challenge. In this chapter I will cover solutions involving environment-responsive cell-penetrating peptides (CPPs). I will discuss properties of CPPs as universal cellular uptake enhancers, and the modifications imparted to CPP-modified nanocarriers to confine CPP activation to diseased tissues.
Collapse
|
10
|
Mechanistic insights into the efficacy of cell penetrating peptide-based cancer vaccines. Cell Mol Life Sci 2018; 75:2887-2896. [PMID: 29508006 PMCID: PMC6061156 DOI: 10.1007/s00018-018-2785-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 11/15/2017] [Accepted: 02/22/2018] [Indexed: 02/07/2023]
Abstract
Immunotherapies are increasingly used to treat cancer, with some outstanding results. Immunotherapy modalities include therapeutic vaccination to eliminate cancer cells through the activation of patient’s immune system against tumor-derived antigens. Nevertheless, the full potential of therapeutic vaccination has yet to be demonstrated clinically because many early generation vaccines elicited low-level immune responses targeting only few tumor antigens. Cell penetrating peptides (CPPs) are highly promising tools to advance the field towards clinical success. CPPs efficiently penetrate cell membranes, even when linked to antigenic cargos, which can induce both CD8 and CD4 T-cell responses. Pre-clinical studies demonstrated that targeting multiple tumor antigens, even those considered to be poorly immunogenic, led to tumor regression. Therefore, CPP-based cancer vaccines represent a flexible and powerful means to extend therapeutic vaccination to many cancer indications. Here, we review recent findings in CPP development and discuss their use in next generation immunotherapies.
Collapse
|
11
|
Lytic EBV infection investigated by detection of Soluble Epstein-Barr virus ZEBRA in the serum of patients with PTLD. Sci Rep 2017; 7:10479. [PMID: 28874674 PMCID: PMC5585268 DOI: 10.1038/s41598-017-09798-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/28/2017] [Indexed: 12/14/2022] Open
Abstract
The ZEBRA protein (encoded by the BZLF1 gene), is the major transcription factor of EBV, expressed upon EBV lytic cycle activation. Several studies highlighted the critical role of EBV lytic infection as a risk factor for lymphoproliferative disorders like post-transplant lymphoproliferative disease (PTLD). Here, we use an antigen-capture ELISA assay specifically designed to detecting the circulating soluble ZEBRA (sZEBRA) in serum samples (threshold value determined at 40ng/mL). We retrospectively investigated a population of 66 transplanted patients comprising 35 PTLD. All the samples from a control population (30 EBV-seronegative subjects and 25 immunocompetent individuals with EBV serological reactivation), classified as sZEBRA < 40ng/mL were assigned as negative. At PTLD diagnosis, EBV genome (quantified by qPCR with EBV DNA>200 copies/mL) and sZEBRA were detectable in 51% and 60% of cases, respectively. In the patients who developed a pathologically-confirmed PTLD, the mean sZEBRA value in cases, was 399 ng/mL +/− 141 versus 53ng/mL +/− 7 in patients who did not (p < 0,001). This is the first report relating to the detection of the circulating ZEBRA in serum specimens, as well as the first analysis dealing with the lytic cycle of EBV in PTLD patients with this new biomarker.
Collapse
|
12
|
Targeted release of transcription factors for human cell reprogramming by ZEBRA cell-penetrating peptide. Int J Pharm 2017. [DOI: 10.1016/j.ijpharm.2017.06.073] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
13
|
Enhancing Antitumor Immune Responses by Optimized Combinations of Cell-penetrating Peptide-based Vaccines and Adjuvants. Mol Ther 2016; 24:1675-85. [PMID: 27377043 DOI: 10.1038/mt.2016.134] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 06/17/2016] [Indexed: 11/08/2022] Open
Abstract
Cell penetrating peptides (CPPs) from the protein ZEBRA are promising candidates to exploit in therapeutic cancer vaccines, since they can transport antigenic cargos into dendritic cells and induce tumor-specific T cells. Employing CPPs for a given cancer indication will require engineering to include relevant tumor-associated epitopes, administration with an appropriate adjuvant, and testing for antitumor immunity. We assessed the importance of structural characteristics, efficiency of in vitro transduction of target cells, and choice of adjuvant in inducing the two key elements in antitumor immunity, CD4 and CD8 T cells, as well as control of tumor growth in vivo. Structural characteristics associated with CPP function varied according to CPP truncations and cargo epitope composition, and correlated with in vitro transduction efficiency. However, subsequent in vivo capacity to induce CD4 and CD8 T cells was not always predicted by in vitro results. We determined that the critical parameter for in vivo efficacy using aggressive mouse tumor models was the choice of adjuvant. Optimal pairing of a particular ZEBRA-CPP sequence and antigenic cargo together with adjuvant induced potent antitumor immunity. Our results highlight the irreplaceable role of in vivo testing of novel vaccine constructs together with adjuvants to select combinations for further development.
Collapse
|
14
|
Derouazi M, Di Berardino-Besson W, Belnoue E, Hoepner S, Walther R, Benkhoucha M, Teta P, Dufour Y, Yacoub Maroun C, Salazar AM, Martinvalet D, Dietrich PY, Walker PR. Novel Cell-Penetrating Peptide-Based Vaccine Induces Robust CD4+ and CD8+ T Cell-Mediated Antitumor Immunity. Cancer Res 2015; 75:3020-31. [PMID: 26116496 DOI: 10.1158/0008-5472.can-14-3017] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 05/02/2015] [Indexed: 12/19/2022]
Abstract
Vaccines that can coordinately induce multi-epitope T cell-mediated immunity, T helper functions, and immunologic memory may offer effective tools for cancer immunotherapy. Here, we report the development of a new class of recombinant protein cancer vaccines that deliver different CD8(+) and CD4(+) T-cell epitopes presented by MHC class I and class II alleles, respectively. In these vaccines, the recombinant protein is fused with Z12, a novel cell-penetrating peptide that promotes efficient protein loading into the antigen-processing machinery of dendritic cells. Z12 elicited an integrated and multi-epitopic immune response with persistent effector T cells. Therapy with Z12-formulated vaccines prolonged survival in three robust tumor models, with the longest survival in an orthotopic model of aggressive brain cancer. Analysis of the tumor sites showed antigen-specific T-cell accumulation with favorable modulation of the balance of the immune infiltrate. Taken together, the results offered a preclinical proof of concept for the use of Z12-formulated vaccines as a versatile platform for the development of effective cancer vaccines.
Collapse
Affiliation(s)
- Madiha Derouazi
- Geneva University Hospitals and University of Geneva, Centre of Oncology, Geneva, Switzerland.
| | | | | | - Sabine Hoepner
- Geneva University Hospitals and University of Geneva, Centre of Oncology, Geneva, Switzerland
| | - Romy Walther
- University of Toulouse, CNRS 5273, UMR STROMALab, Toulouse, France
| | - Mahdia Benkhoucha
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Patrick Teta
- Geneva University Hospitals and University of Geneva, Centre of Oncology, Geneva, Switzerland
| | - Yannick Dufour
- Geneva University Hospitals and University of Geneva, Centre of Oncology, Geneva, Switzerland
| | - Céline Yacoub Maroun
- Geneva University Hospitals and University of Geneva, Centre of Oncology, Geneva, Switzerland
| | | | - Denis Martinvalet
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Pierre-Yves Dietrich
- Geneva University Hospitals and University of Geneva, Centre of Oncology, Geneva, Switzerland
| | - Paul R Walker
- Geneva University Hospitals and University of Geneva, Centre of Oncology, Geneva, Switzerland.
| |
Collapse
|
15
|
Marchione R, Daydé D, Lenormand JL, Cornet M. Vectorisation de molécules biologiques dans Candida albicans grâce au peptide de transduction MD-ZEBRA. J Mycol Med 2015. [DOI: 10.1016/j.mycmed.2015.02.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
16
|
Yoo H, Jung H, Kim SA, Mok H. Multivalent comb-type aptamer-siRNA conjugates for efficient and selective intracellular delivery. Chem Commun (Camb) 2015; 50:6765-7. [PMID: 24830507 DOI: 10.1039/c4cc01620c] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this study, a simple and efficient strategy for selective intracellular delivery of RNA therapeutics into target cancer cells was designed using direct complementary base pairing between chemically conjugated multimeric antisense strands and aptamer-incorporating sense strands.
Collapse
Affiliation(s)
- Hyundong Yoo
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 143-701, Republic of Korea.
| | | | | | | |
Collapse
|
17
|
MD11-mediated delivery of recombinant eIF3f induces melanoma and colorectal carcinoma cell death. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2015; 2:14056. [PMID: 26052528 PMCID: PMC4448995 DOI: 10.1038/mtm.2014.56] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 10/08/2014] [Accepted: 10/17/2014] [Indexed: 11/24/2022]
Abstract
The f subunit of the eukaryotic initiation factor 3 (eIF3f) is downregulated in several cancers and in particular in melanoma and pancreatic cancer cells. Its enforced expression by transient gene transfection negatively regulates cancer cell growth by activating apoptosis. With the aim to increase the intracellular level of eIF3f proteins and activate apoptosis in cancer cell lines, we developed a protein transfer system composed of a cell-penetrating peptide sequence fused to eIF3f protein sequence (MD11-eIF3f). To determine whether exogenously administered eIF3f proteins were able to compensate the loss of endogenous eIF3f and induce cancer cell death, we analyzed the therapeutic action of MD11-eIF3f in several tumor cells. We identified four cell lines respondent to eIF3f-treatment and we evaluated the antitumor properties of the recombinant proteins using dose- and time-dependent studies. Our results demonstrate that this protein delivery approach represents an innovative and powerful strategy for cancer treatment.
Collapse
|
18
|
Marchione R, Daydé D, Lenormand JL, Cornet M. ZEBRA cell-penetrating peptide as an efficient delivery system inCandida albicans. Biotechnol J 2014; 9:1088-94. [DOI: 10.1002/biot.201300505] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 11/28/2013] [Accepted: 01/17/2014] [Indexed: 11/10/2022]
|
19
|
Park K, Yang JA, Lee MY, Lee H, Hahn SK. Reducible hyaluronic acid-siRNA conjugate for target specific gene silencing. Bioconjug Chem 2013; 24:1201-9. [PMID: 23731084 DOI: 10.1021/bc4001257] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Despite wide applications of polymer-drug conjugates, there are only a few polymer-siRNA conjugates like poly(ethylene glycol) conjugated siRNA. In this work, reducible hyaluronic acid (HA)-siRNA conjugate was successfully developed for target specific systemic delivery of siRNA to the liver. The conjugation of siRNA to HA made it possible to form a compact nanocomplex of siRNA with relatively nontoxic linear polyethyleneimine (LPEI). After characterization of HA-siRNA conjugate by size exclusion chromatography (SEC) and gel electrophoresis, its complex formation with LPEI was investigated with a particle analyzer. The HA-siRNA/LPEI complex had a mean particle size of ca. 250 nm and a negative or neutral surface charge at physiological condition. The reducible HA-siRNA/LPEI complex showed a higher in vitro gene silencing efficiency than noncleavable HA-siRNA/LPEI complex. Furthermore, after systemic delivery, apolipoprotein B (ApoB) specific HA-siApoB/LPEI complex was target specifically delivered to the liver, which resulted in statistically significant reduction of ApoB mRNA expression in a dose dependent manner. The HA-siRNA conjugate can be effectively applied as a model system to the treatment of liver diseases using various siRNAs and relatively nontoxic polycations.
Collapse
Affiliation(s)
- Kitae Park
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), San 31 Hyoja-dong, Nam-gu, Pohang, Kyungbuk 790-784, Korea
| | | | | | | | | |
Collapse
|
20
|
Erazo-Oliveras A, Muthukrishnan N, Baker R, Wang TY, Pellois JP. Improving the endosomal escape of cell-penetrating peptides and their cargos: strategies and challenges. Pharmaceuticals (Basel) 2012; 5:1177-1209. [PMID: 24223492 PMCID: PMC3816665 DOI: 10.3390/ph5111177] [Citation(s) in RCA: 313] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 10/25/2012] [Accepted: 10/26/2012] [Indexed: 12/13/2022] Open
Abstract
Cell penetrating peptides (CPPs) can deliver cell-impermeable therapeutic cargos into cells. In particular, CPP-cargo conjugates tend to accumulate inside cells by endocytosis. However, they often remain trapped inside endocytic organelles and fail to reach the cytosolic space of cells efficiently. In this review, the evidence for CPP-mediated endosomal escape is discussed. In addition, several strategies that have been utilized to enhance the endosomal escape of CPP-cargos are described. The recent development of branched systems that display multiple copies of a CPP is presented. The use of viral or synthetic peptides that can disrupt the endosomal membrane upon activation by the low pH of endosomes is also discussed. Finally, we survey how CPPs labeled with chromophores can be used in combination with light to stimulate endosomal lysis. The mechanisms and challenges associated with these intracellular delivery methodologies are discussed.
Collapse
Affiliation(s)
| | | | | | | | - Jean-Philippe Pellois
- Author to whom correspondence should be addressed; ; Tel.: +1-979-845-0101; Fax: +1-979-862-4718
| |
Collapse
|
21
|
Lee TY, Park YS, Garcia GA, Sunahara RK, Woods JH, Yang VC. Cell permeable cocaine esterases constructed by chemical conjugation and genetic recombination. Mol Pharm 2012; 9:1361-73. [PMID: 22404344 DOI: 10.1021/mp200623w] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Cocaine esterase (CocE) is the most efficient cocaine-metabolizing enzyme tested in vivo to date, displaying a rapid clearance of cocaine and a robust protection against cocaine's toxicity. Two potential obstacles to the clinical application of CocE, however, lie in its proteolytic degradation and induced immune response. To minimize these potential obstacles, we attempted nondisruptive cell encapsulation by creating a cell permeable form of CocE, which was achieved by covalently linking a thermally stable CocE mutant (dmCocE) with cell penetrating peptides (CPPs). Two types of CPPs, Tat and the low molecular weight protamine (LMWP), were used in this study. Two types of disulfide-bridged chemical conjugates, Tat-S-S-dmCocE and LMWP-S-S-dmCocE, were synthesized and then purified by heparin affinity chromatography. In addition, four recombinant CPP-dmCocE fusion proteins, Tat-N-dmCocE, LMWP-N-dmCocE, dmCocE-C-Tat, and dmCocE-C-LMWP, were constructed, expressed in Escherichia coli, and purified as soluble proteins. Among these six CPP-dmCocE variants, LMWP-S-S-dmCocE showed the highest cocaine-hydrolyzing activity, and dmCocE-C-Tat had the highest production yield. To evaluate their cellular uptake behavior, a covalently linked fluorophore (FITC) was utilized to visualize the cellular uptake of all six CPP-dmCocE variants in living HeLa cells. All the six variants exhibited cellular uptake, but their intracellular distribution phenotypes differed. While the chemical conjugates showed primarily cytoplasmic distribution, which was likely due to the reduction of the disulfide linkage between CPP and dmCocE, all the other four recombinant fusion proteins displayed both nuclear and cytoplasmic localization, with dmCocE-C-CPP exhibiting higher cytoplasmic distribution during cellular uptake. Based on a balanced consideration of essentials for clinical application, including parameters such as high cocaine-hydrolyzing efficiency, large production yield, major cytoplasmic distribution, etc., the dmCocE-C-Tat fusion protein seems to be the best candidate from this investigation. Further in vivo studies of the cell-encapsulated dmCocE-C-Tat in hydrolyzing cocaine and alleviating immunogenicity and proteolytic degradation in established, clinically relevant mouse models are currently underway in our laboratories. Findings from this research are not only useful for developing other new CPP-CocE constructs but also valuable for establishing a nondisruptive cell-encapsulation technology for other protein therapeutics that are known to be immunogenic for direct clinical application.
Collapse
Affiliation(s)
- Tien-Yi Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109-1065, United States
| | | | | | | | | | | |
Collapse
|
22
|
Greiner VJ, Shvadchak V, Fritz J, Arntz Y, Didier P, Frisch B, Boudier C, Mély Y, de Rocquigny H. Characterization of the mechanisms of HIV-1 Vpr(52–96) internalization in cells. Biochimie 2011; 93:1647-58. [DOI: 10.1016/j.biochi.2011.05.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Accepted: 05/31/2011] [Indexed: 02/08/2023]
|
23
|
Luther EM, Koehler Y, Diendorf J, Epple M, Dringen R. Accumulation of silver nanoparticles by cultured primary brain astrocytes. NANOTECHNOLOGY 2011; 22:375101. [PMID: 21852719 DOI: 10.1088/0957-4484/22/37/375101] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Silver nanoparticles (AgNP) are components of various food industry products and are frequently used for medical equipment and materials. Although such particles enter the vertebrate brain, little is known on their biocompatibility for brain cells. To study the consequences of an AgNP exposure of brain cells we have treated astrocyte-rich primary cultures with polyvinylpyrrolidone (PVP)-coated AgNP. The incubation of cultured astrocytes with micromolar concentrations of AgNP for up to 24 h resulted in a time- and concentration-dependent accumulation of silver, but did not compromise the cell viability nor lower the cellular glutathione content. In contrast, the incubation of astrocytes for 4 h with identical amounts of silver as AgNO(3) already severely compromised the cell viability and completely deprived the cells of glutathione. The accumulation of AgNP by astrocytes was proportional to the concentration of AgNP applied and significantly lowered by about 30% in the presence of the endocytosis inhibitors chloroquine or amiloride. Incubation at 4 °C reduced the accumulation of AgNP by 80% compared to the values obtained for cells that had been exposed to AgNP at 37 °C. These data demonstrate that viable cultured brain astrocytes efficiently accumulate PVP-coated AgNP in a temperature-dependent process that most likely involves endocytotic pathways.
Collapse
Affiliation(s)
- Eva M Luther
- Center for Biomolecular Interactions Bremen, University of Bremen, PO Box 330440, D-28334 Bremen, Germany
| | | | | | | | | |
Collapse
|
24
|
Lu J, Luo H, Wu H, Lan MS, Tan J, Lu D. Recombinant MafA protein containing its own protein transduction domain stimulates insulin gene expression in IEC-6 cells. Life Sci 2011; 89:72-7. [DOI: 10.1016/j.lfs.2011.04.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Revised: 03/21/2011] [Accepted: 04/05/2011] [Indexed: 11/17/2022]
|